Abstract
Studies of the major hemoglobin disorders, β-thalassemia and sickle cell disease (SCD), have laid a foundation for molecular medicine. While enormous progress has been made in understanding gene structure and regulation, translating molecular insights to therapy for the many individuals affected with these disorders has been challenging. Advances in three activities have recently converged to bring novel genetic and potentially curative treatments to clinical trials. First, improved lentiviral vectors for gene transfer into hematopoietic stem cells have revived somatic gene therapy for blood disorders. Second, elucidation of regulatory factors and mechanisms that control the normal developmental switch from fetal to adult hemoglobin has provided a route to reactivation of the fetal form for therapy. Third, revolutionary methods of gene engineering permit molecular insights to be leveraged for patients. Here I review how the promise of molecular medicine to bring transformative treatments to the clinical arena is finally being realized.
eTOC
Orkin discusses how recent curative genetic therapies for β-thalassemia and sickle cell disease are the result of improvements in lentiviral vectors, a detailed understanding of mechanisms that regulate the switch from fetal to adult hemoglobin, and the development of CRISPR/Cas9 gene editing.
Molecular medicine can trace its origins to studies of the inherited hemoglobin disorders. More than seventy years ago, well before elucidation of the structure of DNA or the genetic code, Pauling reported a change in composition of the β-globin chain of the adult hemoglobin tetramer (α2β2) in red cells of individuals with sickle cell anemia, heralding the disorder as the first “molecular disease”1. With development of recombinant DNA methods in the late 1970s, the human globin genes were among the first mammalian genes to be cloned and then fully analyzed by DNA sequencing2,3. At the same time, Kan and Dozy reported a restriction enzyme site polymorphism linked to a β-globin gene harboring the sickle cell mutation, representing the first clinically relevant disease association made through DNA analysis4, and a primitive harbinger of contemporary genome-wide association studies (GWAS). Comprehensive discovery of the diverse mutations in β-thalassemias, inherited disorders of β-chain production, in the 1980s provided the first nearly complete description of a disease at the molecular level, and a critical database for effective DNA-based prenatal diagnosis5.
Research focused on globin genes and developing red blood cells has informed concepts of gene structure and expression, chromatin dynamics, and the role of transcription factors in lineage determination and differentiation. The major cis-regulatory element for expression of the β-globin genes, an extended enhancer known as the locus control region (LCR)6,7, was the first experimentally established super-enhancer8. The looping of the LCR to downstream globin genes has formed the basis for current views of gene activation by enhancers 9,10 and the higher order structure of chromatin and its interactions. Study of the master erythroid transcription factor GATA1 revealed how red cell gene expression is programmed 11,12, and led to the identification of rare inherited red cell and platelet disorders, as well as an underlying somatic mutation characteristic of preleukemia and myeloid leukemia in the setting of trisomy 2113,14.
While these discoveries illustrate the extraordinary power of contemporary molecular methods to dissect genetic diseases and underlying mechanisms, patients rightfully ask “what have you done for us?” Research grant applications promise translation of genetic discoveries to patients, but the sobering reality is that the route to the clinic and positive clinical benefit is challenging and often not straightforward. The early history of genetic therapy for hemoglobin disorders mirrors the development of somatic gene therapy for other inherited conditions: slower than originally anticipated and marred by egregious investigator misjudgments. Soon after the cloning of globin gene sequences and prior to any understanding of how the genes are regulated, globin cDNAs in a plasmid with a selectable marker were transfected into bone marrow cells, which were then infused into patients in a human experiment that became known as the “Cline affair”15. Fortunately, in the intervening years other investigators pursued evidence-based approaches in a decades long path to rational and effective therapy.
The Hemoglobin Disorders
Although β-thalassemia and sickle cell disease (SCD) are granted “orphan” disease status for the purpose of encouraging development of new treatments, they are by no means rare disorders. Together, hundreds of thousands, or millions, of individuals worldwide are affected with two β-thalassemia or sickle (βS) alleles. In sub-Saharan Africa, ~300,000 or more children are born with SCD each year and sadly face >75% mortality by age five largely due to infectious disease. In the United States, the prevalence of SCD is 75–100,000 individuals with an annual health expenditure of ~$1B. The global disease burden of hemoglobin disorders is therefore huge, and anticipated to increase in the coming years16,17.
The β-thalassemias, which result from deficiency (β+) or complete failure (β0) in production of the β-globin chain of adult hemoglobin (α2β2), are due to diverse mutations in the β-globin gene affecting its transcription, RNA processing, or translation of its mRNA into protein5,18. Although SCD is due to a single amino chain in the β-chain (G>V at position 6), which renders sickle hemoglobin (α2βS2) prone to polymerize at low oxygen, the spectrum of clinical severity is notoriously broad. Management of patients with β-thalassemia and SCD has traditionally been largely supportive. For β-thalassemia, red cell transfusion and iron chelation are mainstays of therapy. In SCD, management consists of pain control and penicillin prophylaxis for functional asplenia, supplemented by chronic transfusions for those with intractable pain, stroke, or splenic sequestration. As general medical care in resource-poor countries improves, a marked increase in disease prevalence will further tax health systems. Standard of care for SCD includes oral treatment with hydroxyurea (HU), the only agent with proven substantial clinical benefit19,20. Replacement of the hematopoietic system by marrow transplantation is curative in both β- thalassemia and SCD, and limited by availability of histocompatible donors, the inherent toxicity of myeloablative preconditioning of the recipient, and the overall resource intensive nature of the procedure.
Addressing the primary problem in the hemoglobinopathies
The pathophysiology of the disorders is well understood18,21. In β-thalassemia, deficient or absent β-globin production establishes an imbalance between α-globin and β-globin chains, leading to precipitation of a-globin, hemolysis, and ineffective erythropoiesis. In SCD, polymerization of HbS impairs developing red cells, leading to loss of red cell deformability, microvascular damage, anemia, and the secondary complications of pain and stroke. While therapies may be directed to downstream effects of chain imbalance and sickling, it is axiomatic that targeting the hemoglobin composition should be the most direct and effective means of lessening disease severity and forestalling complications. Well before the contemporary molecular era, astute clinical investigators provided clues to possible solutions. The wide range of severity of the hemoglobinopathies points to the contribution of genetic modifiers. The two strongest modifiers are α-thalassemia (decreased α-globin production) and the level of fetal hemoglobin (HbF). In β-thalassemia, α-thalassemia reduces globin chain imbalance and subsequent hemolysis and ineffective erythropoiesis, despite lowering of the total hemoglobin concentration in the red cell. Since sickling of HbS is concentration-dependent, α-thalassemia is also beneficial in SCD22. Increased HbF replaces missing HbA in β-thalassemia, and in SCD elevated HbF has a two-fold benefit. First, HbF blocks HbS polymerization and second, βS- globin production is reduced concomitantly with increased γ-globin expression due to the reciprocal nature in which the linked genes are transcribed in the human β-like globin complex. The salutary effect of HbF on SCD was first deduced by the pediatrician Janet Watson in 194923. She observed that children with SCD were asymptomatic in the first few months of life and hypothesized that HbF was protective.
Progress in genetic approaches to the hemoglobin disorders that are likely curative has accelerated greatly in recent years, largely due to development of improved vectors for gene transfer into hematopoietic stem cells (HSCs), the advent of transformative gene editing technologies24,25, and a deeper understanding of globin gene regulation.
Globin gene regulation and the fetal-to-adult switch
The organization and expression of human globin genes has been a topic of intense investigation since the molecular cloning of the α- and β-globin gene clusters in bacteriophages in 19782,3. This landmark achievement in the laboratory of Tom Maniatis was reported at the first Hemoglobin Switching Conference, a biannual meeting organized by Arthur Nienhuis and the late George Stamatoyannopoulos, and supported by the NIH, that has fostered development of the field to the present26. The name given to the conference was prescient as it embodied a conviction that understanding how globin genes are regulated would have clinical impact. As the major hemoglobin disorders are due to β-globin gene mutations, only the human β-globin complex on the short arm of chromosome 11 will be discussed further in this Perspective.
Within the β-globin cluster, the β-like genes are arranged downstream of the LCR in the order in which they are expressed during development: LCR- embryonic (ε ) – the duplicated fetal (Gγ–Aγ) –adult (β) —genes (Figure 1) Activation of transcription of each gene is achieved through physical looping and contact with the LCR. The first globin switch, embryonic ε– to fetal γ-globin occurs in the transition to fetal liver hematopoiesis in the second trimester. Thereafter, the critical fetal-adult switch takes place gradually and is completed only in the first postnatal months. In the adult, the level of HbF is ~1%, which is largely restricted to a small population termed F-cells27.
Figure 1. The fetal-to-adult hemoglobin switch.

The critical transition from HbF(α2γ2) to HbA (α2β2) entails a transcriptional switch from engagement of the locus control region (LCR) with the γ-globin genes to the β-globin gene. Two repressor proteins BCL11A and LCR (ZBTB7A) initiate the switch by blocking γ-globin transcription and preventing access by the LCR. Repression of γ-globin gene expression allows interaction of β-globin gene with the LCR and transcription activation.
In adults, the level of HbF is under genetic control of two kinds. Common genetic variation leads to quite modest changes (few %) in HbF. Rare genetic variation, deletions of DNA within the cluster or specific single nucleotide changes in the γ-globin promoters, is associated with substantial (10–30x normal) HbF production, termed hereditary persistence of fetal hemoglobin (HPFH)28(Figure 2) The point mutations in the γ-promoter exert profound effects, such that the level of expression of the linked γ-globin gene is expressed at full level. Therefore, inheritance of an HPFH allele in trans to either a βS or β-thalassemia allele greatly counteracts either chain imbalance or sickling, and may eliminate disease manifestations.
Figure 2. Mechanism of γ-globin gene repression.

The sequence of the γ-globin gene promoter is depicted. The DNA-binding motifs of the two repressors LRF/ZBTB7A and BCL11A are highlighted in blue and red, respectively, with base substitutions observed in patients with HPFH noted below the sequence. Both BCL11A and LRF physically interact with the multicomponent NuRD complex. Binding of BCL11A and NF-Y, an activator that binds a CCAAT box (underlined in yellow) that overlaps a proximal BCL11A motif, oppose each other’s action. BCL11A displaces NF-Y by steric hindrance in the promoter. The boxed region above the sequence indicates a 13bp deletion observed in HPFH and also generated frequently upon CRISPR/Cas9 editing of the promoter 68. Figure modified from Orkin and Bauer 55.
Expression of lineage restricted genes in developing red cell precursors is under the control of the major erythroid transcription factors (TFs), including the master regulator GATA1 and additional factors (TAL1, NF-E2, KLF1)29. None of these TFs exhibit stage-selective expression or activity that can account for the silencing of γ-globin expression in the fetal-adult switch. The search for candidate TFs controlling the switch proved elusive.
Application of GWAS to identify candidate loci regulating HbF level shined new light on the problem. Cotemporaneous studies by Menzel and Thein29 and by Uda and associates30 revealed that a previously unsuspected locus on the short arm of chromosome 2, BCL11A, was strongly associated with F-cell number and HbF level, respectively. SNPs near or within the BCL11A gene reflect common genetic variation that is widely distributed among populations, i.e. they are not restricted to those groups in which β-thalassemia or SCD are prevalent. In concert with other SNPs in the vicinity of the c-Myb locus and within the β-globin gene cluster, such common variation may explain as much as 25–50% of genetic variation of HbF30,31, which is enormous in relation to the majority of genetic contributions attributed to GWAS in most disease settings.
BCL11A has served as an entry point into the mechanistic details of the hemoglobin switch. Consistent with a postulated role as a repressor of γ-globin expression, BCL11A is not expressed as RNA or protein at the embryonic stage of erythroid development, and first appears at the time of fetal liver erythropoiesis in the definitive (or adult) lineage of erythroid cells32. Within human fetal liver erythroid cells, BCL11A mRNA translation is partially blocked by RNA- binding proteins that are subsequently down-regulated at the adult stage33. Down-regulation of BCL11A by short-hairpin RNA (shRNA) in adult erythroid cells (derived from CD34+ stem/progenitor cells) leads to increased HbF, compatible with the proposed action of BCL11A as a γ-globin gene repressor32. In retrospect, given the myriad roles that many TFs play in erythroid cell differentiation and the effects of many parameters, including cell proliferation and maturation, on the relative level of HbF in such cultured cells, these early experiments might only be accepted as successive evidence in favor of BCL11A as a potential therapeutic target.
A priori, silencing of γ-globin gene transcription might rely on a few, or numerous, stage- selective factors. The number of factors involved has direct implications for any target-based reactivation of HbF. If a single factor (or just a couple) is critical, a target-based strategy might be feasible. On the other hand, if many factors are involved, prospects for such therapeutic approaches would be remote. Fortuitously, the former situation appears to apply to control of HbF, as manipulation of BCL11A alone is sufficient to achieve quite high level derepression. The rescue of hematologic parameters of engineered sickle cell mice by erythroid-specific knockout of BCL11A provided crucial insight34. Engineered sickle cell mice lack mouse globin genes and express human globin genes, including the βS-gene, from transgenes35,36. Knockout of BCL11A in the erythroid lineage led to nearly complete rescue of red cell parameters, and pancellular reactivation of HbF to ~30% of total Hb. Thus, removal of the single factor was sufficient to achieve phenotypic rescue. Given that the human globin genes in engineered mice mimic but do not fully recapitulate the normal organization of globin gene clusters, it is not possible to predict whether ~30% HbF constitutes an upper limit for reactivation in a human erythroid cell lacking BCL11A or a lower limit. Nonetheless, the engineered sickle cell mouse experiments provided a persuasive rationale for pursuit of BCL11A as a genetic target for the hemoglobin disorders.
HbF levels are quite sensitive to the expression of BCL11A (Figure 3). Common genetic variation detected in GWAS is associated with a modest (~40% estimated) reduction in BCL11A expression and a small increase in HbF37. Moreover, in patients with the autism-like BCL11A haploinsufficiency syndrome HbF levels average ~10–15% with a wide range38–40. Highly associated SNPs lie within the second intron of the BCL11A gene, within and surrounding, an ~10 kb erythroid-specific enhancer element characterized by 3 subregions (+55, +58, +62) marked by DNase I hypersensitivity37. A causal SNP in the +63 HS disrupts a canonical GATA-TAL1 binding site. Despite the large size of the BCL11A erythroid enhancer, a GATA1 binding site within +58 confers a major portion of activity to the entire enhancer41. This “Achilles heel” represents an especially favorable target for genetic disruption by either zinc-finger nucleases or CRISPR/Cas941–43. Besides the marked reduction in BCL11A expression observed upon targeting these sequences, the effects of the edits on expression are entirely restricted to erythroid cells44. Cell-selectivity is important as BCL11A is expressed and critical for proper hematopoietic stem cell function and B-lymphoid development, though the relative thresholds for impairment in these cells are unknown45,46. For example, in haploinsufficient individuals, B-cell development and overall hematopoiesis appear normal38.
Figure 3. Genetics of BCL11A and HbF output.

The consequences of genetic alterations at the BCL11A locus for HbF expression are summarized. The panels depicting common low and high HbF genotypes at the BCL11A locus illustrate the effect of common genetic variation, as measured in GWAS, on BCL11A expression and HbF expression. Natural variants at BCL11A lie within the erythroid-specific enhancer of the BCL11A gene and determine the level of transcription and output of BCL11A protein. The “high HbF” allele is associated with reduced BCL11A protein and a modest increase in HbF. Deletion of the erythroid-specific enhancer nearly abolishes BCL11A expression, leading to marked increase in HbF, as shown in the third panel. Therapeutic gene editing of the erythroid enhancer, specifically at the “Achilles heel” region, greatly reduces BCL11A expression but not to the extent of the enhancer deletion. Nonetheless, the level of HbF expression is significantly increased and is sufficient to ameliorate the severity of either β-thalassemia or SCD. Figure modified from Hardison and Blobel 87.
Although potent and essential as a regulator of Hb silencing, BCL11A is not the sole factor. A second factor, named ZBTB7A or LRF, is comparable in its activity, as shown by knockout in immortalized CD34+ derived erythroid cells (HUDEP-2)47. Knockout of either BCL11A or LRF leads to ~40–50% HbF in this model. HbF levels are 90–100% in cells with combined knockout. Both factors physically associate with the NuRD corepressor complex, which confers both remodeling and histone deacetylase activities47,48. CRISPR/Cas9 mutagenesis of the various subunits of NuRD demonstrates that specific paralogs of each subunit are required for HbF repression49. Further support for a cofactor role for the NuRD complex in HbF repression is provided by recent findings that ZNF410, a pentadactyl DNA binding protein, exhibits remarkable specificity for activation of the CHD4 locus, which encodes its ATP-dependent remodeling subunit50,51.
Recent studies provide an increasingly detailed view of how γ-globin gene repression is controlled (Figure 2). Whereas chromosomal looping between the LCR and each downstream globin gene is believed to be required for transcriptional activation, the choice between expression of γ- and β-globin genes appears to be executed locally. Specifically, γ-globin gene repression depends on the two repressors (BCL11A and LRF), each binding to a cognate recognition site within the γ-globin promoter52,53. Three closely spaced zinc-fingers at the C-terminus of BCL11A specific recognition of a unit motif, TGACCA, which is duplicated in γ-globin promoter. Remarkably, BCL11A binds selectively to the more distal of the duplicated sites, and some of the rare, naturally occurring point mutations leading to the HPFH syndrome disrupt the TGACCA motif53. The duplicated TGACCA motifs overlap duplicated CCAAT boxes, recognition sequences for the ubiquitous CCAAT-binding protein complex NF-Y. Of note, in cells expressing γ-globin, NF-Y selectively binds the more proximal CCAAT motif54. Detailed functional studies indicate that BCL11A binding to its cognate TGACCA site displaces NF-Y from its proximal site to initiate repression. Recruitment of the NuRD complex by BCL11A is believed to stabilize repression. LRF recognizes a GC-rich motif at a more distal position in the γ-globin promoter, which is also the site of rare single base changes in HPFH52. As LRF also associates with the NuRD protein complex, it is likely that concerted recruitment of NuRD to the promoter is required for stable repression54. Such a concerted action may explain how loss of either BCL11A or LRF leads to a similar deficit in repression, whereas combined loss further augments repression.
Strategies for genetic management of hemoglobin disorders
Disease pathophysiology can be reversed only by addressing the quality and quantity of hemoglobin within red cells. In β-thalassemias, raising the level of β-like globin (either β- or γ-globin) is the goal. As the relative imbalance of α- and β-globin chains is reduced, the extent of ineffective erythropoiesis and anemia is lessened. Approaches to genetic therapy include correcting the specific mutation for the given thalassemia allele(s), expression of an exogenously introduced β-like globin gene, or reactivation of endogenous γ-globin to produce high levels of HbF55. In principle, correction of the underlying sickle mutation is the simplest approach as the same base change is present in all βS alleles. Expression of a non-sickling β-globin or γ-globin, or reactivation of endogenous γ-globin, is more tractable with current technology.
Transcriptional output from the human β-globin cluster is directed by the LCR, which physically loops to downstream globin genes for activation. As such, expression from the γ- and β-globin genes is reciprocal. In fetal erythroid cells, the LCR loops to the γ-globin genes, and β-globin transcription is largely prevented. Binding of BCL11A and LRF to the γ-globin promoters down-regulates transcription, and frees up the LCR (by unknown mechanisms) to loop to the β-globin gene. Reactivation of γ-globin expression upon removal of BCL11A (or LRF) leads to concomitant down-regulation of β-globin expression. As a practical consequence of these mechanisms, reactivation of the endogenous γ-globin genes in SCD reduces HbS production. In effect, increased HbF affords two benefits in SCD: first, HbF impairs the sickling process and second, HbS production itself is turned down. Similarly, in β-thalassemia, increased expression of endogenous HbF replaces missing HbA (from β-globin deficiency) and down-regulates β-globin production which lessens chain imbalance and ineffective erythropoiesis.
The following strategies are available for genetic therapy of the hemoglobin disorders (Table 1):
Globin gene addition: introduction of a vector that provides for expression of an exogenous globin (either β-globin, γ-globin, or a non-sickling β-globin)
Gene correction: introduction of gene modifying reagents to correct the underlying gene mutation
Reactivation of endogenous HbF: introduction of vector or gene modifying reagents that lead to re-expression of γ-globin genes in adult erythroid cells due to either down-regulation of a repressor (BCL11A) or alteration of a repressor binding site (i.e. recreation of an HPFH allele).
Table 1. Targets for gene manipulation therapy.
Proposed targets for genetic treatment of β-thalassemia and SCD are presented with the corresponding method, approach, and desired outcome. HDR: homology directed repair; NHEJ: non-homologous end-joining.
| Target | Method | Approach | Outcome | HbF expression | Clinical trial |
|---|---|---|---|---|---|
| β-globin gene | CRISPR/Cas9 (or similar) | HDR | Corrected gene | No change | |
| BCL11A mRNA | Lentivirus | shRNA-mediated erythroid knockdown | Reduced BCL11A expression | Reactivation | Yes |
| BCL11A erythroid enhancer | CRISPR/Cas9 (or other editing) | NHEJ | Reduced BCL11A expression | Reactivation | Yes |
| BCL11A binding site in γ-globin gene promoters | CRISPR/Cas9 (or other editing) | NHEJ | Mimic HPFH allele | Reactivation | |
| BCL11A binding site in γ-globin gene promoters | Base editing | C>T conversion | Mimic HPFH allele | Reactivation |
Genetic technologies
At present, genetic therapy for hematopoietic disorders requires ex vivo modification of enriched stem/progenitor cells (CD34+ cells) and preconditioning of the host with chemotherapy to ablate endogenous hematopoiesis. For a durable result, gene transfer into long-term repopulating hematopoietic stem cells (HSCs) is critical. Early studies in gene therapy employed retroviral vectors. Inefficient transduction of HSCs, shutoff or inconsistent vector expression, and insertional mutagenesis plagued early clinical trials56. Transition to use of lentiviral vectors, which exhibit more random insertion into the genome and stable expression, has greatly accelerated progress in the field.
Gene modifying strategies have undergone a revolution in recent years24,57. Zinc-fingers and TALENs allowed “proof-of-principle” experiments for gene disruption by non-homologous end joining (NHEJ) and by homology-directed repair (HDR). Indeed, knockout of the HIV CCR5 receptor in lymphoid cells with zinc-fingers constituted the first human gene editing trial58. The advent of CRISPR/Cas9 editing has brought gene editing to the fore, given its ease of use, flexibility, high efficiency and low off-target action. The newer method of base editing expands options for gene manipulation. As discussed below, critical issues in clinical application include efficiency, off target effects, and the nature of the target under consideration for therapeutic intervention.
Clinical trials for hemoglobinopathies (Table 2)
Table 2. Gene therapy approaches in clinical development for hemoglobinopathies.
Data are derived from recent publications and ClinicalTrials.gov as of November 2020.
| Molecular approach | Disorder | Clinicaltrials.gov identifier | Sponsoring institution | Approval |
|---|---|---|---|---|
| Globin addition (modified β) Lentivirus |
SCD β-thalassemia |
NCT02140554 NCT03207009 NCT02906202 |
Bluebird bio | ZYNTEGLO® (Europe) |
| Globin addition (modified β) Lentivirus |
SCD | NCT02247843 | UCLA | |
| Globin addition (modified γ) Lentivirus |
SCD | NCT02186418 | Cincinnati Children’s Hospital Medical Center | |
| HbF reactivation Erythroid shRNA knockdown of BCL11A Lentivirus |
SCD | NCT03282656 | Boston Children’s Hospital/Bluebird bio | |
| HbF reactivation Gene editing of BCL11A enhancer CRISPR/Cas9 |
β-thalassemia SCD |
NCT03655678 (Climb THAL-111) NCT03745287 (Climb SCD-121) |
CRISPR Therapeutics/Vertex |
Globin gene addition:
The first promising clinical trials for hemoglobinopathies have relied on lentiviral vectors in which a β-globin gene is expressed under the control of elements derived from the β-globin cluster LCR. After a long period of vector development, lentiviruses that efficiently transduce HSPCs and express β-globin were identified. In 2010 LeBoulch and colleagues reported a single patient with transfusion-dependent βE/β0-thalassemia, the most common form of β-thalassemia in Southeast Asia, that was treated with a lentivirus expressing a mutated β-globin βT87Q with anti-sickling properties59. Following allogeneic hematopoietic reconstitution with modified HSPCs, the patient became transfusion-independent and maintained a Hb of 9–10 g/dl, of which ~30% was derived from the vector. The majority of exogenously expressed globin was derived from a dominant, myeloid-based clone, which was expanded due to insertional HMGA2 gene activation in erythroid cells. Clonal dominance was lost by 12 years post therapy and associated with the need for intermittent transfusions60.
The largest clinical experience in globin addition therapy is that reported by Cavazzana and colleagues in which 22 patients with transfusion-dependent β-thalassemia were treated with LentiGlobin BB305, a vector that was slightly modified from the used in the earlier LeBoulch patient60. Patients with β+-thalassemia (i.e. those with at least one non-β0-allele) fared significantly better than β0-thalassemia individuals, of which only 3/9 became transfusion-independent. Clinical benefit was documented as transfusion requirements were reduced by 73% overall with associated improvement in hematologic parameters. No serious adverse events were reported. Despite the positive findings of this trial, full correction of pathophysiology was not achieved. LentiGlobin BB305 has been approved in Europe as treatment for β+-thalassemia patients >12 year of age. In a separate trial (TIGET, BTHAL, NCT02453477), which is no longer recruiting patients, 3 adults and 6 children with β-thalassemia were treated with a β-globin lentivirus (GLOCE) by intrabone administration of transduced HSCs61. Transfusion requirements were reduced but not eliminated in the adults and 3 of 4 evaluable pediatric patients became transfusion independent.
LentiGlobin BB305 has also been used in a similarly designed trial to treat patients with SCD. A single patient has been described with correction of hallmarks of the disease62. A recent update of the trial, now including a total of 14 SCD patients, described resolution of sickle crises in almost all patients63.
BCL11A-directed reactivation of HbF:
The alternative strategy of increasing endogenous HbF to replace missing HbA in β-thalassemia or retard sickling in SCD has the added theoretical benefit of leveraging the reciprocal pattern of expression of the γ- and β-globin genes within the globin cluster to reduce transcription of the mutant β-gene. The most critical parameter influencing the potential success of this approach is the level to which HbF can be raised with an intervention. Based on preclinical studies, near complete loss of BCL11A in erythroid precursors would likely reactivate HbF to ~40–50% of total Hb in a given modified cell, a level well within a therapeutic range for either β-thalassemia or SCD. Recreation of a single HPFH mutation of the “Greek”-HPFH variety would be similarly beneficial.
Lentiviral shRNA to BCL11A:
Based on preclinical studies showing that either erythroid-specific knockout of BCL11A or erythroid-directed lentiviral shRNA directed to BCL11A reactivates HbF to levels sufficient to rescue SCD mice46,64, a clinical trial was designed and initiated at Boston Children’s Hospital. The trial is innovative in several respects. It is the first trial in which an shRNA, in this instance was embedded in a miRNA scaffold (a shmiR), has been employed. Second, the shmiR was contained within a lentiviral vector similar to that used in the globin addition trials in order to limit expression to erythroid precursors. Third, the trial represents the first in man to assess reactivation of endogenous HbF. The initial results of the trial, which has recently been reported65, are remarkable for the level of HbF reactivation observed and for the overall consistency patient to patient. Of 6 patients 6–21 months post therapy, F-cell level were ~70% with F/F-cell = 9.6–13.8 pg (33–47% of total Hb/cell) with a total HbF of 22–40%. All patients became symptom-free and no serious adverse events were observed. Hematological parameters indicated some residual ongoing hemolysis but very much reduced from pre-therapy values. HbF was not detected in ~25% of red cells, i.e. that they were the product either of unmodified HSCs or from residual host HSCs that were not ablated in preconditioning. The former possibility is more likely but additional studies are required. The initial findings of this trial are important in that they fully validate the preclinical science underlying the overall strategy.
Gene editing:
Preliminary findings of a collaborative trial between CRISRP Therapeutics and Vertex to reactivate HbF by CRISPR/Cas9-mediated disruption of the “Achilles heel” in the BCL11A enhancer41 have been reported recently66,67. In this instance, Cas9 protein and an sgRNA in an RNP complex was electroporated into HSPCs ex vivo and then reinfused into preconditioned 5 β-thalassemia or 2 SCD patients in the trials to date. The results revealed thus far are very promising. All patients with transfusion-dependent β-thalassemia remained transfusion-free from 2 months post-therapy. The 2 patients with SCD patient experienced no vaso-occlusive crises post-therapy. All treated patients maintained near normal hemoglobin levels and in the two individuals with SCD HbF was >40%. To date, no serious adverse events have been reported. These first-in-human CRISPR/Cas9 editing trials of HSPCs appear to demonstrate clinical benefit of gene editing in the hemoglobin disorders. Additional clinical trials based on gene editing of the BCL11A enhancer are anticipated from other academic and commercial entities.
In development:
At least three other approaches are in preclinical development. First, based on the critical role of the BCL11A binding site in the γ-globin promoters, CRISPR/Cas9 gene editing to disrupt the motif will likely be brought forward as a strategy to recreate an HPFH-like allele in patients. The presence of duplicated sequences in the promoters tends to favor a 13 bp deletion within the promoter or a larger deletion that reduces an allele to a single γ-globin gene 68, although specific gRNAs or choice a different Cas9-like protein permits discrete editing of the motif. Second, the BCL11A binding motif is also favorable for use of base editors that convert C-T24,69. In this setting the site can be mutated to prevent occupancy of BCL11A protein, and therefore mimic the HPFH syndrome. Base editing has a theoretical advantage over gene editing in that base conversion does not involve a cut in the target DNA and therefore may avoid cellular responses to DNA breaks and therefore afford greater safety. Base editing to create other HPFH-like mutations in the γ-globin promoter can also be envisioned. Third, precise correction of the sickle mutation or any given β-thalassemia mutation by HDR is in theory the most direct approach as it converts the target allele to the normal β-globin sequence. To date, attention has focused largely on correction of the sickle mutation using CRISPR/Cas9 and related procedures70,71. The challenges to clinical application of HDR for SCD relate to the overall efficiency at which the correction is achieved and the concomitant generation of indels, which in effect convert the βS gene to a β-thalassemia allele. Improvements in methodology have greatly improved the frequency of gene correction, and therefore it is likely that this approach will be brought to clinical trials in the future72.
Lessons from clinical trials
Several lessons are emerging at this early stage of clinical translation:
First, current technology --either lentiviral gene therapy or gene editing-- is capable of achieving substantial improvement in hematological parameters and clinical benefit in patients with β-thalassemia and SCD. To be sure, further studies are required to assess durability, consistency of responses, and safety of current approaches. Nonetheless, the demonstration that transfusion requirements can be alleviated in β-thalassemia and sickle crises reduced or eliminated in SCD constitutes a significant milestone in genetic therapy. With a goal of complete restoration of normal red cell production and dynamics, there is room for improvement.
Second, achieving meaningful therapeutic benefit necessitates modifying a substantial proportion of the host HSCs. Based on the findings in SCD patients who have exhibited mixed chimerism after bone marrow transplantation from a compatible donor, discussion in the field has centered on what might constitute the lowest threshold for the proportion of gene modified cells for clinical benefit73,74. A threshold of 10–20% has generally been taken as a consensus estimate that a given manipulation of HSPCs ex vivo – gene transduction, gene editing, gene correction by HDR -- might likely be in “therapeutic range”. Preclinical and clinical data thus far argue however that higher levels will be needed to achieve transformative and consistent benefit to patients. In lentiviral globin addition trials, enhanced HSC transduction has been critical in both β-thalassemia and SCD. In preclinical experiments, gene editing frequencies of 70% or greater are required to obtain consistent and robust results in reconstituted mice42.
Third, reactivation of endogenous HbF as a therapeutic strategy for β-thalassemia and SCD, a long-sought goal envisioned decades ago, is feasible and likely to provide results comparable to that of globin gene addition. The in vivo findings in patients are remarkably consistent with preclinical studies involving manipulation of BCL11A34,64,65. Whether the theoretical benefit of greater physiologic control of globin gene expression achieved by reactivation of HbF (as opposed to globin addition) is realized in clinical outcomes remains to be explored. Validation of BCL11A as a therapeutic target in patients also supports a rationale for small molecule-based therapeutics directed to the protein as an alternative, and more scalable, strategy to reduce disease burden in populations.
Challenges going forward
In the short term, the durability and safety of the ongoing gene-based trials need to be assessed carefully. Given the different therapeutic strategies, comparison, one-to-another and head-to-head where possible, will be important in assessing the best options for patients. The relative benefits and risks of the different approaches merit careful consideration. Although the current generation of lentiviral vectors has an increasingly good risk profile, the specter of insertional mutagenesis or oncogene activation can never be entirely eliminated75,76. In principle, transcriptional repression of the inserted lentivirus long after therapy is possible. In trails employing CRISPR-based editing, off-targeting by sgRNAs and larger scale chromosomal alterations and losses must be assessed in the setting of clinical trials77,78. In a true sense, as we enter the realm of therapeutic genome modifications we participate in a game of roulette where the risk of an intervention must be weighed against the morbidity of the target disease in the background of ongoing mutation of the genome that occurs with natural aging79.
As therapies move to approval, the ease and cost of preparation of molecular reagents versus large scale lentivirus production may impact commercial competitiveness. Apart from these important aspects, ex vivo genetic therapy for hemoglobinopathies is auto-transplantation with modified HSPCs, and therefore resource intensive. Innovative ways of reimbursement are needed in order to reduce inequities in availability of novel therapies to patients and yet encourage commercial investment in development of these treatments for “orphan” diseases80.
Chemotherapeutic preconditioning currently employed for myeloablative transplantation, as used in gene therapy, carries potential risks for patients. Non-ablative preconditioning with antibodies or antibody toxin conjugates may offer safer and less toxic alternatives in the future81,82. Instead of HSPCs harvested from patients, HSCs generated from pluripotent stem cells (iPSCs) that have been genetically corrected in culture could in principle constitute an alternative source of cells for hematopoietic reconstitution83. For this goal to be realized, however, major enhancements in derivation of authentic HSCs and their expansion are needed84.
As currently executed, genetic therapy for β-thalassemia and SCD cannot adequately address the true burden of disease either in the US or elsewhere, and particularly in resource-limited areas17. In vivo genetic therapy, in which a viral product or a gene modifying complex is delivered once or repeatedly, is envisioned as a possible approach85,86. Rapid progress has been made in in vivo genetic therapy for eye disorders and prospects for treatment of disorders for which correction of cells in the liver is beneficial are exciting. Given the need to modify HSCs at a substantial level to achieve lessening of disease pathophysiology in thalassemia and SCD, transformative technologies will be needed to realize this vision for blood disorders.
Conclusion
While much remains to be done, advancing genetic therapies to patients with β-thalassemia and SCD once again affirms the inestimable value of long-term investment in fundamental science. Besides the benefit afforded to patients with these disorders, experience in the various approaches to gene transfer and gene modification will accelerate progress toward innovative therapies for other conditions that may be less common and attract fewer resources. The field of molecular medicine is at an inflection point in the development of curative genetic therapies.
Acknowledgments
Work from the author’s laboratory has been supported by the Howard Hughes Medical Institute, grants from the National Institutes of Health, and an award from the Doris Duke Charitable Foundation. The author is an inventor on patents issued pertaining to BCL11A and control of fetal hemoglobin. Non-exclusive licenses have been provided through the Technology & Innovation Development Office (TIDO) at Boston Children’s Hospital.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Pauling L, Itano HA, Singer SJ, Wells IC (1949). Sickle cell anemia a molecular disease. Science. 110, 543–548. [DOI] [PubMed] [Google Scholar]
- 2.Lauer J, Shen CK, Maniatis T (1980). The chromosomal arrangement of human alpha-like globin genes: sequence homology and alpha-globin gene deletions. Cell. 20, 119–130. [DOI] [PubMed] [Google Scholar]
- 3.Lawn RM, Fritsch EF, Parker RC, Blake G, Maniatis T (1978). The isolation and characterization of linked delta- and beta-globin genes from a cloned library of human DNA. Cell. 15, 1157–1174. [DOI] [PubMed] [Google Scholar]
- 4.Kan YW, Dozy AM (1978). Polymorphism of DNA sequence adjacent to human beta-globin structural gene: relationship to sickle mutation. Proc Natl Acad Sci U S A. 75, 5631–5635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Orkin SH, Kazazian HH Jr., Antonarakis SE, Goff SC, Boehm CD, Sexton JP, Waber PG, Giardina PJ (1982). Linkage of beta-thalassaemia mutations and beta-globin gene polymorphisms with DNA polymorphisms in human beta-globin gene cluster. Nature. 296, 627–631. [DOI] [PubMed] [Google Scholar]
- 6.Yagi M, Gelinas R, Elder JT, Peretz M, Papayannopoulou T, Stamatoyannopoulos G, Groudine M (1986). Chromatin structure and developmental expression of the human alpha-globin cluster. Mol Cell Biol. 6, 1108–1116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Grosveld F, van Assendelft GB, Greaves DR, Kollias G (1987). Position-independent, high- level expression of the human beta-globin gene in transgenic mice. Cell. 51, 975–985. [DOI] [PubMed] [Google Scholar]
- 8.Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-Andre V, Sigova AA, Hoke HA, Young RA (2013). Super-enhancers in the control of cell identity and disease. Cell. 155, 934–947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Deng W, Lee J, Wang H, Miller J, Reik A, Gregory PD, Dean A, Blobel GA (2012). Controlling long-range genomic interactions at a native locus by targeted tethering of a looping factor. Cell. 149, 1233–1244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Dekker J (2008). Gene regulation in the third dimension. Science. 319, 1793–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Tsai SF, Martin DI, Zon LI, D’Andrea AD, Wong GG, Orkin SH (1989). Cloning of cDNA for the major DNA-binding protein of the erythroid lineage through expression in mammalian cells. Nature. 339, 446–451. [DOI] [PubMed] [Google Scholar]
- 12.Evans T, Felsenfeld G (1989). The erythroid-specific transcription factor Eryf1: a new finger protein. Cell. 58, 877–885. [DOI] [PubMed] [Google Scholar]
- 13.Nichols KE, Crispino JD, Poncz M, White JG, Orkin SH, Maris JM, Weiss MJ (2000). Familial dyserythropoietic anaemia and thrombocytopenia due to an inherited mutation in GATA1. Nat Genet. 24, 266–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Wechsler J, Greene M, McDevitt MA, Anastasi J, Karp JE, Le Beau MM, Crispino JD (2002). Acquired mutations in GATA1 in the megakaryoblastic leukemia of Down syndrome. Nat Genet. 32, 148–152. [DOI] [PubMed] [Google Scholar]
- 15.Beutler E (2001). The Cline affair. Mol Ther. 4, 396–397. [DOI] [PubMed] [Google Scholar]
- 16.Orkin SH, Higgs DR (2010). Medicine. Sickle cell disease at 100 years. Science. 329, 291–292. [DOI] [PubMed] [Google Scholar]
- 17.Weatherall DJ (2010). The inherited diseases of hemoglobin are an emerging global health burden. Blood. 115, 4331–4336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Benz EJ Jr., Forget BG (1982). The thalassemia syndromes: models for the molecular analysis of human disease. Annu Rev Med. 33, 363–373. [DOI] [PubMed] [Google Scholar]
- 19.Platt OS, Orkin SH, Dover G, Beardsley GP, Miller B, Nathan DG (1984). Hydroxyurea increases fetal hemoglobin production in sickle cell anemia. Trans Assoc Am Physicians. 97, 268–274. [PubMed] [Google Scholar]
- 20.Charache S, Terrin ML, Moore RD, Dover GJ, Barton FB, Eckert SV, McMahon RP, Bonds DR (1995). Effect of hydroxyurea on the frequency of painful crises in sickle cell anemia. Investigators of the Multicenter Study of Hydroxyurea in Sickle Cell Anemia. N Engl J Med. 332, 1317–1322. [DOI] [PubMed] [Google Scholar]
- 21.Nathan DG (1972). Thalassemia. N Engl J Med. 286, 586–594. [DOI] [PubMed] [Google Scholar]
- 22.Coletta M, Hofrichter J, Ferrone FA, Eaton WA (1982). Kinetics of sickle haemoglobin polymerization in single red cells. Nature. 300, 194–197. [DOI] [PubMed] [Google Scholar]
- 23.Watson J (1948). The significance of the paucity of sickle cells in newborn Negro infants. Am J Med Sci. 215, 419–423. [DOI] [PubMed] [Google Scholar]
- 24.Anzalone AV, Koblan LW, Liu DR (2020). Genome editing with CRISPR-Cas nucleases, base editors, transposases and prime editors. Nat Biotechnol. 38, 824–844. [DOI] [PubMed] [Google Scholar]
- 25.Doudna JA, Charpentier E (2014). Genome editing. The new frontier of genome engineering with CRISPR-Cas9. Science. 346, 1258096. [DOI] [PubMed] [Google Scholar]
- 26.Orkin SH (1990). Globin gene regulation and switching: circa 1990. Cell. 63, 665–672. [DOI] [PubMed] [Google Scholar]
- 27.Zago MA, Wood WG, Clegg JB, Weatherall DJ, O’Sullivan M, Gunson H (1979). Genetic control of F cells in human adults. Blood. 53, 977–986. [PubMed] [Google Scholar]
- 28.Forget BG (1998). Molecular basis of hereditary persistence of fetal hemoglobin. Ann N Y Acad Sci. 850, 38–44. [DOI] [PubMed] [Google Scholar]
- 29.Orkin SH, Zon LI (2008). Hematopoiesis: an evolving paradigm for stem cell biology. Cell. 132, 631–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Lettre G, Sankaran VG, Bezerra MA, Araujo AS, Uda M, Sanna S, Cao A, Schlessinger D, Costa FF, Hirschhorn JN, et al. (2008). DNA polymorphisms at the BCL11A, HBS1L-MYB, and beta-globin loci associate with fetal hemoglobin levels and pain crises in sickle cell disease. Proc Natl Acad Sci U S A. 105, 11869–11874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Galarneau G, Palmer CD, Sankaran VG, Orkin SH, Hirschhorn JN, Lettre G (2010). Fine-mapping at three loci known to affect fetal hemoglobin levels explains additional genetic variation. Nat Genet. 42, 1049–1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sankaran VG, Menne TF, Xu J, Akie TE, Lettre G, Van Handel B, Mikkola HK, Hirschhorn JN, Cantor AB, Orkin SH (2008). Human fetal hemoglobin expression is regulated by the developmental stage-specific repressor BCL11A. Science. 322, 1839–1842. [DOI] [PubMed] [Google Scholar]
- 33.Basak A, Munschauer M, Lareau CA, Montbleau KE, Ulirsch JC, Hartigan CR, Schenone M, Lian J, Wang Y, Huang Y, et al. (2020). Control of human hemoglobin switching by LIN28B-mediated regulation of BCL11A translation. Nat Genet. 52, 138–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Xu J, Peng C, Sankaran VG, Shao Z, Esrick EB, Chong BG, Ippolito GC, Fujiwara Y, Ebert BL, Tucker PW, et al. (2011). Correction of sickle cell disease in adult mice by interference with fetal hemoglobin silencing. Science. 334, 993–996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Ryan TM, Townes TM, Reilly MP, Asakura T, Palmiter RD, Brinster RL, Behringer RR (1990). Human sickle hemoglobin in transgenic mice. Science. 247, 566–568. [DOI] [PubMed] [Google Scholar]
- 36.Paszty C, Brion CM, Manci E, Witkowska HE, Stevens ME, Mohandas N, Rubin EM (1997). Transgenic knockout mice with exclusively human sickle hemoglobin and sickle cell disease. Science. 278, 876–878. [DOI] [PubMed] [Google Scholar]
- 37.Bauer DE, Kamran SC, Lessard S, Xu J, Fujiwara Y, Lin C, Shao Z, Canver MC, Smith EC, Pinello L, et al. (2013). An erythroid enhancer of BCL11A subject to genetic variation determines fetal hemoglobin level. Science. 342, 253–257.24115442 [Google Scholar]
- 38.Basak A, Hancarova M, Ulirsch JC, Balci TB, Trkova M, Pelisek M, Vlckova M, Muzikova K, Cermak J, Trka J, et al. (2015). BCL11A deletions result in fetal hemoglobin persistence and neurodevelopmental alterations. J Clin Invest. 125, 2363–2368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Levy J, Coussement A, Dupont C, Guimiot F, Baumann C, Viot G, Passemard S, Capri Y, Drunat S, Verloes A, et al. (2017). Molecular and clinical delineation of 2p15p16.1 microdeletion syndrome. Am J Med Genet A. 173, 2081–2087. [DOI] [PubMed] [Google Scholar]
- 40.Mimouni-Bloch A, Yeshaya J, Kahana S, Maya I, Basel-Vanagaite L (2015). A de-novo interstitial microduplication involving 2p16.1-p15 and mirroring 2p16.1-p15 microdeletion syndrome: Clinical and molecular analysis. Eur J Paediatr Neurol. 19, 711–715. [DOI] [PubMed] [Google Scholar]
- 41.Canver MC, Smith EC, Sher F, Pinello L, Sanjana NE, Shalem O, Chen DD, Schupp PG, Vinjamur DS, Garcia SP, et al. (2015). BCL11A enhancer dissection by Cas9-mediated in situ saturating mutagenesis. Nature. 527, 192–197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wu Y, Zeng J, Roscoe BP, Liu P, Yao Q, Lazzarotto CR, Clement K, Cole MA, Luk K, Baricordi C, et al. (2019). Highly efficient therapeutic gene editing of human hematopoietic stem cells. Nat Med. 25, 776–783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Vierstra J, Reik A, Chang KH, Stehling-Sun S, Zhou Y, Hinkley SJ, Paschon DE, Zhang L, Psatha N, Bendana YR, et al. (2015). Functional footprinting of regulatory DNA. Nat Methods. 12, 927–930. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Smith EC, Luc S, Croney DM, Woodworth MB, Greig LC, Fujiwara Y, Nguyen M, Sher F, Macklis JD, Bauer DE, et al. (2016). Strict in vivo specificity of the Bcl11a erythroid enhancer. Blood. 128, 2338–2342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Luc S, Huang J, McEldoon JL, Somuncular E, Li D, Rhodes C, Mamoor S, Hou S, Xu J, Orkin SH (2016). Bcl11a Deficiency Leads to Hematopoietic Stem Cell Defects with an Aging-like Phenotype. Cell Rep. 16, 3181–3194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Guda S, Brendel C, Renella R, Du P, Bauer DE, Canver MC, Grenier JK, Grimson AW, Kamran SC, Thornton J, et al. (2015). miRNA-embedded shRNAs for Lineage-specific BCL11A Knockdown and Hemoglobin F Induction. Mol Ther. 23, 1465–1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Masuda T, Wang X, Maeda M, Canver MC, Sher F, Funnell AP, Fisher C, Suciu M, Martyn GE, Norton LJ, et al. (2016). Transcription factors LRF and BCL11A independently repress expression of fetal hemoglobin. Science. 351, 285–289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Xu J, Bauer DE, Kerenyi MA, Vo TD, Hou S, Hsu YJ, Yao H, Trowbridge JJ, Mandel G, Orkin SH (2013). Corepressor-dependent silencing of fetal hemoglobin expression by BCL11A. Proc Natl Acad Sci U S A. 110, 6518–6523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Sher F, Hossain M, Seruggia D, Schoonenberg VAC, Yao Q, Cifani P, Dassama LMK, Cole MA, Ren C, Vinjamur DS, et al. (2019). Rational targeting of a NuRD subcomplex guided by comprehensive in situ mutagenesis. Nat Genet. 51, 1149–1159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Vinjamur DS, Yao Q, Cole MA, McGuckin C, Ren C, Zeng J, Hossain M, Pinello L, Bauer DE (2020). ZNF410 represses fetal globin by devoted control of CHD4/NuRD. Blood. https://ash.confex.com/ash/2020/webprogram/Paper142134.html. [DOI] [PMC free article] [PubMed]
- 51.Lan X, ren R, Feng R, Ly LC, Lan Y, Zhang Z, Aboreden N, Qin K, Horton JR, Grevet JD, et al. (2020). ZNF410 uniquely activates the NuRD component CHD4 to silence fetal hemoglobin expression. Molecular Cell. DOI: 10.1016/j.molcel.2020.1011.1006. [DOI] [PMC free article] [PubMed]
- 52.Martyn GE, Wienert B, Yang L, Shah M, Norton LJ, Burdach J, Kurita R, Nakamura Y, Pearson RCM, Funnell APW, et al. (2018). Natural regulatory mutations elevate the fetal globin gene via disruption of BCL11A or ZBTB7A binding. Nat Genet. 50, 498–503. [DOI] [PubMed] [Google Scholar]
- 53.Liu N, Hargreaves VV, Zhu Q, Kurland JV, Hong J, Kim W, Sher F, Macias-Trevino C, Rogers JM, Kurita R, et al. (2018). Direct Promoter Repression by BCL11A Controls the Fetal to Adult Hemoglobin Switch. Cell. 173, 430–442 e417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Liu N, Xu S, Yao Q, Kai Y, Hsu JY, Sakon P, Pinello L, Yuan GC, Bauer DE, Orkin SH (2020). Transcription factor competition at the g-globin promoters controls hemoglobin switching. Nature Genetics. in press. [DOI] [PMC free article] [PubMed]
- 55.Orkin SH, Bauer DE (2019). Emerging Genetic Therapy for Sickle Cell Disease. Annu Rev Med. 70, 257–271. [DOI] [PubMed] [Google Scholar]
- 56.Baum C, Dullmann J, Li Z, Fehse B, Meyer J, Williams DA, von Kalle C (2003). Side effects of retroviral gene transfer into hematopoietic stem cells. Blood. 101, 2099–2114. [DOI] [PubMed] [Google Scholar]
- 57.Urnov FD, Rebar EJ, Holmes MC, Zhang HS, Gregory PD (2010). Genome editing with engineered zinc finger nucleases. Nat Rev Genet. 11, 636–646. [DOI] [PubMed] [Google Scholar]
- 58.Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin MA, Nichol G, et al. (2014). Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 370, 901–910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Cavazzana-Calvo M, Payen E, Negre O, Wang G, Hehir K, Fusil F, Down J, Denaro M, Brady T, Westerman K, et al. (2010). Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature. 467, 318–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Thompson AA, Walters MC, Kwiatkowski J, Rasko JEJ, Ribeil JA, Hongeng S, Magrin E, Schiller GJ, Payen E, Semeraro M, et al. (2018). Gene Therapy in Patients with Transfusion-Dependent beta-Thalassemia. N Engl J Med. 378, 1479–1493. [DOI] [PubMed] [Google Scholar]
- 61.Marktel S, Scaramuzza S, Cicalese MP, Giglio F, Galimberti S, Lidonnici MR, Calbi V, Assanelli A, Bernardo ME, Rossi C, et al. (2019). Intrabone hematopoietic stem cell gene therapy for adult and pediatric patients affected by transfusion-dependent ss-thalassemia. Nat Med. 25, 234–241. [DOI] [PubMed] [Google Scholar]
- 62.Ribeil JA, Hacein-Bey-Abina S, Payen E, Magnani A, Semeraro M, Magrin E, Caccavelli L, Neven B, Bourget P, El Nemer W, et al. (2017). Gene Therapy in a Patient with Sickle Cell Disease. N Engl J Med. 376, 848–855. [DOI] [PubMed] [Google Scholar]
- 63.Thompson AA, Walters MC, Mapara MY, Kwiatkowski J, Kirshnamurti L, Aygun B, Kasow KA, Rifkin-Zenenberg S, Schmidt M, DelCarpini J, et al. (2020). Resolution of serious vaso-opcclusive pain crises and reduction in patient-reported pain intensity: results from the ongoing phase 1/2 HBG-206 Group C study of LentiGlobin for sickle cell disease (bb111) gene therapy. Blood. 136 (supplement, https://ash.confex.com/ash/2020/webprogram/Paper134940.html. [Google Scholar]
- 64.Brendel C, Guda S, Renella R, Bauer DE, Canver MC, Kim YJ, Heeney MM, Klatt D, Fogel J, Milsom MD, et al. (2016). Lineage-specific BCL11A knockdown circumvents toxicities and reverses sickle phenotype. J Clin Invest. 126, 3868–3878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Esrick EB, Lehmann LE, Biffi A, Achebe M, Brendel C, Ciuculescu MF, Daley H, MacKinnon B, Morris E, Federica A, et al. (2020). Post-translational genetic silencing of BCL11A to treat sickle cell disease. NEJM. DOI: 10.1056/NEJMoa2029392, [DOI] [PMC free article] [PubMed]
- 66.Corbacioglu S, Cappelini MD, Chapin J, Chu-Osier N, Fernandez CM, Foell J, de la Fuente J, Grupp S, Ho TW, Kattamis A, et al. (2020). Initial safety and efficacy results with a single dose of autologous CRISPR-CAS9 modified CD34+ hemtopoietic stem and progenitor cells in transfusion dependent b-thalassemia and sickle cell disease. HemaSphere. 4, S1. [Google Scholar]
- 67.Frangoul H, Bobruff Y, Cappelini MD, Corbacioglu S, Fernandez CM, de la Fuente J, Grupp S, Handgretinger R, Ho TW, Imren S, et al. (2020). Safety and efficacy of CTX001 in patients with transfusion-dependent b-thalassemia and sickle cell disease: early results from the Climb THAL-111 and Climb SCD-121 studies of autologous CRISPR-CAS9-modified CD34+ hematopoietic stem and progenitor cells. Blood. 136 (supplement), https://ash.confex.com/ash/2020/webprogram/Paper139575. [Google Scholar]
- 68.Traxler EA, Yao Y, Wang YD, Woodard KJ, Kurita R, Nakamura Y, Hughes JR, Hardison RC, Blobel GA, Li C, et al. (2016). A genome-editing strategy to treat beta- hemoglobinopathies that recapitulates a mutation associated with a benign genetic condition. Nat Med. 22, 987–990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zeng J, Wu Y, Ren C, Bonanno J, Shen AH, Shea D, Gehrke JM, Clement K, Luk K, Yao Q, et al. (2020). Therapeutic base editing of human hematopoietic stem cells. Nat Med. 26, 535–541. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Dever DP, Bak RO, Reinisch A, Camarena J, Washington G, Nicolas CE, Pavel-Dinu M, Saxena N, Wilkens AB, Mantri S, et al. (2016). CRISPR/Cas9 beta-globin gene targeting in human haematopoietic stem cells. Nature. 539, 384–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Martin RM, Ikeda K, Cromer MK, Uchida N, Nishimura T, Romano R, Tong AJ, Lemgart VT, Camarena J, Pavel-Dinu M, et al. (2019). Highly Efficient and Marker-free Genome Editing of Human Pluripotent Stem Cells by CRISPR-Cas9 RNP and AAV6 Donor- Mediated Homologous Recombination. Cell Stem Cell. 24, 821–828 e825. [DOI] [PubMed] [Google Scholar]
- 72.Anzalone AV, Randolph PB, Davis JR, Sousa AA, Koblan LW, Levy JM, Chen PJ, Wilson C, Newby GA, Raguram A, et al. (2019). Search-and-replace genome editing without double-strand breaks or donor DNA. Nature. 576, 149–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Walters MC, Patience M, Leisenring W, Rogers ZR, Aquino VM, Buchanan GR, Roberts IA, Yeager AM, Hsu L, Adamkiewicz T, et al. (2001). Stable mixed hematopoietic chimerism after bone marrow transplantation for sickle cell anemia. Biol Blood Marrow Transplant. 7, 665–673. [DOI] [PubMed] [Google Scholar]
- 74.Wu CJ, Gladwin M, Tisdale J, Hsieh M, Law T, Biernacki M, Rogers S, Wang X, Walters M, Zahrieh D, et al. (2007). Mixed haematopoietic chimerism for sickle cell disease prevents intravascular haemolysis. Br J Haematol. 139, 504–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, et al. (2008). Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 118, 3132–3142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Williams DA, Thrasher AJ (2014). Concise review: lessons learned from clinical trials of gene therapy in monogenic immunodeficiency diseases. Stem Cells Transl Med. 3, 636–642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Cullot G, Boutin J, Toutain J, Prat F, Pennamen P, Rooryck C, Teichmann M, Rousseau E, Lamrissi-Garcia I, Guyonnet-Duperat V, et al. (2019). CRISPR-Cas9 genome editing induces megabase-scale chromosomal truncations. Nat Commun. 10, 1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Zuccaro MV, Xu J, Mitchell C, Marin D, Zimmerman R, Rana B, Weinstein E, King RT, Palmerola KL, Smith ME, et al. (2020). Allele-Specific Chromosome Removal after Cas9 Cleavage in Human Embryos. Cell. [DOI] [PubMed]
- 79.Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, Lindsley RC, Mermel CH, Burtt N, Chavez A, et al. (2014). Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med. 371, 2488–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Orkin SH, Reilly P (2016). MEDICINE. Paying for future success in gene therapy. Science. 352, 1059–1061. [DOI] [PubMed] [Google Scholar]
- 81.Czechowicz A, Kraft D, Weissman IL, Bhattacharya D (2007). Efficient transplantation via antibody-based clearance of hematopoietic stem cell niches. Science. 318, 1296–1299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Czechowicz A, Palchaudhuri R, Scheck A, Hu Y, Hoggatt J, Saez B, Pang WW, Mansour MK, Tate TA, Chan YY, et al. (2019). Selective hematopoietic stem cell ablation using CD117-antibody-drug-conjugates enables safe and effective transplantation with immunity preservation. Nat Commun. 10, 617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hanna J, Wernig M, Markoulaki S, Sun CW, Meissner A, Cassady JP, Beard C, Brambrink T, Wu LC, Townes TM, et al. (2007). Treatment of sickle cell anemia mouse model with iPS cells generated from autologous skin. Science. 318, 1920–1923. [DOI] [PubMed] [Google Scholar]
- 84.Vo LT, Daley GQ (2015). De novo generation of HSCs from somatic and pluripotent stem cell sources. Blood. 125, 2641–2648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cohen J, Kaiser J (2019). Gates and NIH join forces on HIV and sickle cell diseases. Science. 366, 558–559. [DOI] [PubMed] [Google Scholar]
- 86.Wang H, Georgakopoulou A, Psatha N, Li C, Capsali C, Samal HB, Anagnostopoulos A, Ehrhardt A, Izsvak Z, Papayannopoulou T, et al. (2019). In vivo hematopoietic stem cell gene therapy ameliorates murine thalassemia intermedia. J Clin Invest. 129, 598–615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hardison RC, Blobel GA (2013). Genetics. GWAS to therapy by genome edits? Science. 342, 206–207.24115432 [Google Scholar]
